Network


Latest external collaboration on country level. Dive into details by clicking on the dots.

Hotspot


Dive into the research topics where Shi-chang Deng is active.

Publication


Featured researches published by Shi-chang Deng.


PLOS ONE | 2013

MiR-130b Is a Prognostic Marker and Inhibits Cell Proliferation and Invasion in Pancreatic Cancer through Targeting STAT3

Gang Zhao; Jun-gang Zhang; Ying Shi; Qi Qin; Yang Liu; Bo Wang; Kui Tian; Shi-chang Deng; Xiang Li; Shuai Zhu; Qiong Gong; Yi Niu; Chunyou Wang

Accumulating evidence indicates that microRNAs (miRNAs) are aberrantly expressed in human cancer and contribute to the tumorigenesis, but their roles in pancreatic cancer are still largely unknown. In this study, our data showed that miR-130b was significantly downregulated in 52 pairs of pancreatic cancer tissues and five cell lines. Furthermore, the deregulated miR-130b was correlated with worse prognosis, increased tumor size, late TNM stage, lymphatic invasion and distant metastasis. Multivariate analysis showed that miR-130b expression was a significant and independent prognostic predictor for pancreatic cancer patients. Functional studies indicated that the overexpression of miR-130b dramatically suppressed the proliferation of pancreatic cancer cells both in vitro and in vivo, which could be attributed to the induction of apoptosis and cell cycle arrest at S phase. Meanwhile, an overexpressed miR-130b remarkably inhibited the invasive ability of pancreatic cancer cells. Moreover, the dual luciferase assay revealed that STAT3 was directly targeted by miR-130b, which was further confirmed by the inverse expression of miR-130b and STAT3 in pancreatic cancer samples. Our findings suggested that miR-130b might have a considerable potential in prognosis identification and application of therapy for pancreatic cancer.


Molecular Cancer Therapeutics | 2013

miRNA-141, Downregulated in Pancreatic Cancer, Inhibits Cell Proliferation and Invasion by Directly Targeting MAP4K4

Gang Zhao; Bo Wang; Yang Liu; Jun-gang Zhang; Shi-chang Deng; Qi Qin; Kui Tian; Xiang Li; Shuai Zhu; Yi Niu; Qiong Gong; Chunyou Wang

miRNAs are associated with various types of cancer due to their ability to affect expression of genes that modulate tumorigenesis. In this study, we explored the role of miR-141 in pancreatic cancer. The analysis of clinical characteristics showed that miR-141 was significantly downregulated in tissues and cell lines of pancreatic cancer. Moreover, the decreased miR-141 level was significantly associated with tumor size and TNM stage, as well as lymph node and distant metastasis. Meanwhile, both Kaplan–Meier and multivariate survival analysis showed decreased miR-141 were associated with overall survival. Overexpression of miR-141 in pancreatic cancer cells inhibited cell proliferation, clonogenicity, and invasion; induced G1 arrest and apoptosis; and enhanced chemosensitivity. To understand how miR-141 mediates the phenotype of pancreatic cancer cells, a bioinformatics tool was used to identify MAP4K4 as a potential target of miR-141. The Dual-Luciferase reporter gene assay showed that miR-141 binds directly to the 3′-untranslated region (3′UTR) of MAP4K4 to inhibit MAP4K4 expression. Western blot and quantitative real-time PCR (qRT-PCR) analyses revealed that MAP4K4 expression was inversely correlated with miR-141 expression both in pancreatic cancer samples and cell lines. Knockdown of MAP4K4 inhibited cell proliferation, clonogenicity, and invasion, induced G1 arrest and apoptosis, and enhanced chemosensitivity. In a nude mouse xenograft model, both overexpression of miR-141 and knockdown of MAP4K4 significantly repressed pancreatic cancer cell growth. Therefore, we conclude that miR-141 targets MAP4K4, acts as a tumor suppressor in pancreatic cancer cells, and may serve as a novel therapeutic agent for miRNA-based pancreatic cancer therapy. Mol Cancer Ther; 12(11); 2569–80. ©2013 AACR.


Molecular Cancer Therapeutics | 2013

MiR-148b functions as a tumor suppressor in pancreatic cancer by targeting AMPKα1

Gang Zhao; Jun-gang Zhang; Yang Liu; Qi Qin; Bo Wang; Kui Tian; Lin Liu; Xiang Li; Yi Niu; Shi-chang Deng; Chunyou Wang

miRNAs are small noncoding RNAs that participate in a variety of biologic processes, and dysregulation of miRNA is always associated with cancer development and progression. Aberrant expression of miR-148b has been found in some types of cancer, but its expression and potential biologic role in pancreatic cancer are still largely unknown. In this study, our data showed that miR-148b was significantly downregulated in 48 pairs of human pancreatic cancer tissues and five cell lines. Furthermore, the deregulated miR-148b was correlated with increased tumor size, late tumor–node–metastasis stage, lymphatic invasion, distant metastasis, and worse prognosis in pancreatic cancer. Functional studies indicated overexpression of miR-148b dramatically suppressed the growth of cancer cells, attributable to induction of apoptosis and cell-cycle arrest at S-phase. Meanwhile, miR-148b remarkably inhibited invasion and enhanced chemosensitivity of pancreatic cancer cells. Moreover, ectopic expression of miR-148b was able to inhibit tumorigenicity in nude mice. Further studies revealed that AMPKα1 might be the direct target gene of miR-148b, and overexpressed AMPKα1 inversely correlated with miR-148b in pancreatic cancer. Silencing of AMPKα1 with RNA interference inhibited the growth of pancreatic cancer cells in vitro and in vivo and also induced apoptosis, cell-cycle arrest, and inhibited invasion of cancer cells, which is consistent with the effects of miR-148b overexpression. In conclusion, miR-148b can inhibit cell proliferation, invasion, and enhance chemosensitivity of pancreatic cancer by targeting AMPKα1. Our present results implicate the potential effects of miR-148b on prognosis and treatment of pancreatic cancer. Mol Cancer Ther; 12(1); 83–93. ©2012 AACR.


Cancer Letters | 2014

Chronic pancreatitis and pancreatic cancer demonstrate active epithelial–mesenchymal transition profile, regulated by miR-217-SIRT1 pathway

Shi-chang Deng; Shuai Zhu; Bo Wang; Xiang Li; Yang Liu; Qi Qin; Qiong Gong; Yi Niu; Cheng Xiang; Jingyuan Chen; Jin Yan; Shijiang Deng; Tao Yin; Ming Yang; Heshui Wu; Chunyou Wang; Gang Zhao

Epithelial-mesenchymal transition (EMT) is supposed to be an inflammation induced response which may take a central role in tumorigenesis. Since recent evidence indicates that microRNAs may be involved in EMT, the present study set out to reveal the miRNA which might regulate the EMT in CP (chronic pancreatitis) and PC (pancreatic cancer) and its potential mechanism. Firstly, we provided evidence that both CP and PC tissues demonstrated active EMT profile. Consistently, miR-217 was obviously down-regulated in CP, PC and TGF-β1 treated PC cells, while negatively correlated to its direct target SIRT1. Moreover, either ectopic expression of miR-217 or inhibition of SIRT1 remarkably induced mesenchymal to epithelial transition (MET) in TGF-β1 treated PC cells. On the contrary, miR-217 inhibitor promoted EMT in PC cells but not in SIRT-knockdown PC cells. Clinical information from a cohort of 54 PC patients demonstrated that down-regulated miR-217 was positively correlated with late tumor stage, lymphatic invasion, vascular infiltration and distant metastasis. These results suggest that the overexpressed TGF-β1 in inflammation triggers the deregulation of the miR-217-SIRT1 pathway and then promotes the EMT process, which might be involved in the tumorigenesis of PC. Additionally, miR-217 may function as a novel target and predictor for PC prevention and therapy.


World Journal of Gastroenterology | 2013

Effects of different resuscitation fluid on severe acute pancreatitis

Gang Zhao; Jun-gang Zhang; Heshui Wu; Jin Tao; Qi Qin; Shi-chang Deng; Yang Liu; Lin Liu; Bo Wang; Kui Tian; Xiang Li; Shuai Zhu; Chunyou Wang

AIM To compare effects of different resuscitation fluid on microcirculation, inflammation, intestinal barrier and clinical results in severe acute pancreatitis (SAP). METHODS One hundred and twenty patients with SAP were enrolled at the Pancreatic Disease Institute between January 2007 and March 2010. The patients were randomly treated with normal saline (NS group), combination of normal saline and hydroxyethyl starch (HES) (SH group), combination of normal saline, hydroxyethyl starch and glutamine (SHG group) in resuscitation. The ratio of normal saline to HES in the SH and SHG groups was 3:1. The glutamine (20% glutamine dipeptide, 100 mL/d) was supplemented into the resuscitation liquid in the SHG group. Complications and outcomes including respiratory and abdominal infection, sepsis, abdominal hemorrhage, intra-abdominal hypertension, abdominal compartment syndrome (ACS), renal failure, acute respiratory distress syndrome (ARDS), multiple organ dysfunction syndrome (MODS), operation intervention, length of intensive care unit stay, length of hospital stay, and mortality at 60 d were compared. Moreover, blood oxygen saturation (SpO2), gastric intramucosal pH value (pHi), intra-abdominal pressure (IAP), inflammation cytokines, urine lactulose/mannitol (L/M) ratio, and serum endotoxin were investigated to evaluate the inflammatory reaction and gut barrier. RESULTS Compared to the NS group, patients in the SH and SHG groups accessed the endpoint more quickly (3.9 ± 0.23 d and 4.1 ± 0.21 d vs 5.8 ± 0.25 d, P < 0.05) with less fluid volume (67.26 ± 28.53 mL/kg/d, 61.79 ± 27.61 mL/kg per day vs 85.23 ± 21.27 mL/kg per day, P < 0.05). Compared to the NS group, incidence of renal dysfunction, ARDS, MODS and ACS in the SH and SHG groups was obviously lower. Furthermore, incidence of respiratory and abdominal infection was significantly decreased in the SH and SHG groups, while no significant difference in sepsis was seen. Moreover, less operation time was needed in the SH and SHG group than the NS group, but the difference was not significant. The mortality did not differ significantly among these groups. Blood SpO2 and gastric mucosal pHi in the SH and SHG groups increased more quickly than in the NS group, while IAP was significantly decreased in the SH and SHG group. Moreover, the serum tumor necrosis factor-α, interleukin-8 and C-reactive protein levels in the SH and SHG groups were obviously lower than in the NS group at each time point. Furthermore, urine L/M ratio and serum endotoxin were significantly lower in the SH group and further decreased in the SHG group. CONCLUSION Results indicated that combination of normal saline, HES and glutamine are more efficient in resuscitation of SAP by relieving inflammation and sustaining the intestinal barrier.


Journal of Surgical Oncology | 2010

Mechanical stiffness of liver tissues in relation to integrin β1 expression may influence the development of hepatic cirrhosis and hepatocellular carcinoma.

Gang Zhao; Jing Cui; Qi Qin; Jun-gang Zhang; Lin Liu; Shi-chang Deng; Chenlin Wu; Ming Yang; Shaoshan Li; Chunyou Wang

Increased rigidity of the extracellular matrix (ECM) is commonly associated with hepatocellular carcinoma (HCC). The purpose of this study was to quantitate the mechanical stiffness of various hepatic tissues, evaluate integrin β1 expression, and investigate the correlation between these two factors in the development of HCC.


Oncotarget | 2016

Hypoxia-induced lncRNA-NUTF2P3-001 contributes to tumorigenesis of pancreatic cancer by derepressing the miR-3923/KRAS pathway.

Xiang Li; Shijiang Deng; Shuai Zhu; Yan Jin; Shi-peng Cui; Jingyuan Chen; Cheng Xiang; Qun-ying Li; Chi He; Shufeng Zhao; Hengyu Chen; Yi Niu; Yang Liu; Shi-chang Deng; Chunyou Wang; Gang Zhao

Recent studies indicate that long non-coding RNAs (lncRNAs) play crucial roles in numerous cancers, while their function in pancreatic cancer is rarely elucidated. The present study identifies a functional lncRNA and its potential role in tumorigenesis of pancreatic cancer. Microarray co-assay for lncRNAs and mRNAs demonstrates that lncRNA-NUTF2P3-001 is remarkably overexpressed in pancreatic cancer and chronic pancreatitis tissues, which positively correlates with KRAS mRNA expression. After downregulating lncRNA-NUTF2P3-001, the proliferation and invasion of pancreatic cancer cell are significantly inhibited both in vitro and vivo, accompanying with decreased KRAS expression. The dual-luciferase reporter assay further validates that lncRNA-NUTF2P3-001 and 3′UTR of KRAS mRNA competitively bind with miR-3923. Furthermore, miR-3923 overexpression simulates the inhibiting effects of lncRNA-NUTF2P3-001-siRNA on pancreatic cancer cell, which is rescued by miR-3923 inhibitor. Specifically, the present study further reveals that lncRNA-NUTF2P3-001 is upregulated in pancreatic cancer cells under hypoxia and CoCl2 treatment, which is attributed to the binding of hypoxia-inducible factor-1α (HIF-1α) to hypoxia response elements (HREs) in the upstream of KRAS promoter. Data from pancreatic cancer patients show a positive correlation between lncRNA-NUTF2P3-001 and KRAS, which is associated with advanced tumor stage and worse prognosis. Hence, our data provide a new lncRNA-mediated regulatory mechanism for the tumor oncogene KRAS and implicate that lncRNA-NUTF2P3-001 and miR-3923 can be applied as novel predictors and therapeutic targets for pancreatic cancer.


Pancreatology | 2013

Nicotinamide prohibits proliferation and enhances chemosensitivity of pancreatic cancer cells through deregulating SIRT1 and Ras/Akt pathways

Jun-gang Zhang; Gang Zhao; Qi Qin; Bo Wang; Lin Liu; Yang Liu; Shi-chang Deng; Kui Tian; Chunyou Wang

BACKGROUND Nicotinamide (NAM), the precursor for the synthesis of NAD(+) and also an inhibitor of SIRT1, has been discovered to inhibit some types of cancer. However, little is known about the effects of NAM on pancreatic cancer cells. Since previous research showed that SIRT1 and K-Ras/Akt signaling acted as a promoter in tumorigenesis of pancreatic cancer, our present research set out to explore whether NAM inhibits proliferation and facilitates chemosensitivity in pancreatic cancer cells as well as the potential mechanisms involving SIRT1 and K-Ras/Akt pathway. METHODS Cell viability was assessed by MTT assay, and apoptosis and cell cycle were measured by flow cytometry. Cell invasive ability was evaluated by matrigel invasion assays. The activity of SIRT1 was measured by the Fluor de Lys deacetylation assay. Expression levels of SIRT1, K-Ras, Phosphated Akt (P-Akt, Ser-473) and Akt were measured using western blot. In vivo tumor growth was performed in pancreatic cancer cells xenografts. RESULTS NAM inhibited the proliferation of pancreatic cancer cells in a dose-dependent manner, and significantly induced apoptosis and cell cycle arrest in G2/M phase. Moreover, NAM obviously restrained cell invasive ability and increased the chemosensitivity. NAM significantly inhibited the activity of SIRT1 and decreased expression of SIRT1, K-Ras and P-Akt. Further, NAM prohibited proliferation and enhanced GEM antitumor activity in vivo. CONCLUSIONS Our results implied that NAM might be a potential therapeutic agent for human pancreatic cancer treatment through downregulating SIRT1, K-Ras and P-Akt expression.


Oncotarget | 2015

MiR-652 inhibits acidic microenvironment-induced epithelial-mesenchymal transition of pancreatic cancer cells by targeting ZEB1

Shi-chang Deng; Xiang Li; Yi Niu; Shuai Zhu; Yan Jin; Shijiang Deng; Jingyuan Chen; Yang Liu; Chi He; Tao Yin; Zhiyong Yang; Jing Tao; Jiongxin Xiong; Heshui Wu; Chunyou Wang; Gang Zhao

Recent evidences suggest that the acidic microenvironment might facilitate epithelial mesenchymal transition (EMT) of tumor cells, while the effects of acidity on EMT of pancreatic cancer (PC) remain undefined. The present study demonstrated that acidity suppressed miR-652 expression, which further promoted EMT process by absenting inhibition on the transcriptional factor ZEB1 expression. At first, we found that acidity remarkably enhanced invasion ability of PC cells accompanying with increased mesenchymal and decreased epithelial markers. Meanwhile, miRNAs-microarray showed that miR-652, the potential regulator of ZEB1, was distinctly decreased in acidity-treated PC cells. Furthermore, restoration of miR-652 reversed acidity-induced EMT by inhibiting ZEB1 expression, while miR-652 inhibitor induced EMT in normal PC cells through promoting ZEB1 expression. Nevertheless, knockdown of ZEB1 significantly suppressed acidity-induced EMT in PC cells, but ZEB1 overexpression rescued the EMT which was inhibited by miR-652 overexpression. The in vivo results showed that the tumor growth and liver metastasis were remarkably retarded by both miR-652 overexpression and ZEB1 knockdown. The clinical samples further revealed that miR-652 was decreased in PC tissues and antagonistically correlated with ZEB1 expression, associating with late tumor stage, lymphatic invasion and metastasis. In conclusion, our study indicated a novel acidity/miR-652/ZEB1/EMT axis in the tumorigenesis of PC.


Cell Death and Disease | 2017

ASIC1 and ASIC3 contribute to acidity-induced EMT of pancreatic cancer through activating Ca2|[plus]||[sol]|RhoA pathway

Shuai Zhu; Hai-Yun Zhou; Shi-chang Deng; Shijiang Deng; Chi He; Xiang Li; Jingyuan Chen; Yan Jin; Zhuang-Li Hu; Fang Wang; Chunyou Wang; Gang Zhao

Extracellular acid can have important effects on cancer cells. Acid-sensing ion channels (ASICs), which emerged as key receptors for extracellular acidic pH, are differently expressed during various diseases and have been implicated in underlying pathogenesis. This study reports that ASIC1 and ASIC3 are mainly expressed on membrane of pancreatic cancer cells and upregulated in pancreatic cancer tissues. ASIC1 and ASIC3 are responsible for an acidity-induced inward current, which is required for elevation of intracellular Ca2+ concentration ([Ca2+]i). Inhibition of ASIC1 and ASIC3 with siRNA or pharmacological inhibitor significantly decreased [Ca2+]i and its downstream RhoA during acidity and, thus, suppressed acidity-induced epithelial–mesenchymal transition (EMT) of pancreatic cancer cells. Meanwhile, downregulating [Ca2+]i with calcium chelating agent BAPTA-AM or knockdown of RhoA with siRNA also significantly repressed acidity-induced EMT of pancreatic cancer cells. Significantly, although without obvious effect on proliferation, knockdown of ASIC1 and ASIC3 in pancreatic cancer cells significantly suppresses liver and lung metastasis in xenograft model. In addition, ASIC1 and ASIC3 are positively correlated with expression of mesenchymal marker vimentin, but inversely correlated with epithelial marker E-cadherin in pancreatic cancer cells. In conclusion, this study indicates that ASICs are master regulator of acidity-induced EMT. In addition, the data demonstrate a functional link between ASICs and [Ca2+]i/RhoA pathway, which contributes to the acidity-induced EMT.

Collaboration


Dive into the Shi-chang Deng's collaboration.

Top Co-Authors

Avatar

Chunyou Wang

Huazhong University of Science and Technology

View shared research outputs
Top Co-Authors

Avatar

Gang Zhao

Huazhong University of Science and Technology

View shared research outputs
Top Co-Authors

Avatar

Shuai Zhu

Huazhong University of Science and Technology

View shared research outputs
Top Co-Authors

Avatar

Xiang Li

Huazhong University of Science and Technology

View shared research outputs
Top Co-Authors

Avatar

Yang Liu

Huazhong University of Science and Technology

View shared research outputs
Top Co-Authors

Avatar

Qi Qin

Huazhong University of Science and Technology

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Yi Niu

Huazhong University of Science and Technology

View shared research outputs
Top Co-Authors

Avatar

Bo Wang

Huazhong University of Science and Technology

View shared research outputs
Researchain Logo
Decentralizing Knowledge